Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jeannette L. Bennicelli is active.

Publication


Featured researches published by Jeannette L. Bennicelli.


The New England Journal of Medicine | 2008

Safety and Efficacy of Gene Transfer for Leber’s Congenital Amaurosis

Albert M. Maguire; Francesca Simonelli; Eric A. Pierce; Edward N. Pugh; Federico Mingozzi; Jeannette L. Bennicelli; Sandro Banfi; Kathleen Marshall; Francesco Testa; Enrico Maria Surace; Settimio Rossi; Arkady Lyubarsky; Valder R. Arruda; Barbara A. Konkle; Edwin M. Stone; Junwei Sun; Jonathan B. Jacobs; L. F. Dell'Osso; Richard W. Hertle; Jian Xing Ma; T. Michael Redmond; Xiaosong Zhu; Bernd Hauck; Olga Zelenaia; Kenneth S. Shindler; Maureen G. Maguire; J. Fraser Wright; Nicholas J. Volpe; Jennifer Wellman McDonnell; Alberto Auricchio

Lebers congenital amaurosis (LCA) is a group of inherited blinding diseases with onset during childhood. One form of the disease, LCA2, is caused by mutations in the retinal pigment epithelium-specific 65-kDa protein gene (RPE65). We investigated the safety of subretinal delivery of a recombinant adeno-associated virus (AAV) carrying RPE65 complementary DNA (cDNA) (ClinicalTrials.gov number, NCT00516477 [ClinicalTrials.gov]). Three patients with LCA2 had an acceptable local and systemic adverse-event profile after delivery of AAV2.hRPE65v2. Each patient had a modest improvement in measures of retinal function on subjective tests of visual acuity. In one patient, an asymptomatic macular hole developed, and although the occurrence was considered to be an adverse event, the patient had some return of retinal function. Although the follow-up was very short and normal vision was not achieved, this study provides the basis for further gene therapy studies in patients with LCA.


The Lancet | 2009

Age-dependent effects of RPE65 gene therapy for Leber's congenital amaurosis: a phase 1 dose-escalation trial

Albert M. Maguire; Katherine A. High; Alberto Auricchio; J. Fraser Wright; Eric A. Pierce; Francesco Testa; Federico Mingozzi; Jeannette L. Bennicelli; Gui-shuang Ying; Settimio Rossi; Ann Fulton; Kathleen Marshall; Sandro Banfi; Daniel C. Chung; Jessica I. W. Morgan; Bernd Hauck; Olga Zelenaia; Xiaosong Zhu; Leslie Raffini; Frauke Coppieters; Elfride De Baere; Kenneth S. Shindler; Nicholas J. Volpe; Enrico Maria Surace; Carmela Acerra; Arkady Lyubarsky; T. Michael Redmond; Edwin M. Stone; Junwei Sun; Jenni Fer Uvellman Mcdonnell

BACKGROUND Gene therapy has the potential to reverse disease or prevent further deterioration of vision in patients with incurable inherited retinal degeneration. We therefore did a phase 1 trial to assess the effect of gene therapy on retinal and visual function in children and adults with Lebers congenital amaurosis. METHODS We assessed the retinal and visual function in 12 patients (aged 8-44 years) with RPE65-associated Lebers congenital amaurosis given one subretinal injection of adeno-associated virus (AAV) containing a gene encoding a protein needed for the isomerohydrolase activity of the retinal pigment epithelium (AAV2-hRPE65v2) in the worst eye at low (1.5 x 10(10) vector genomes), medium (4.8 x 10(10) vector genomes), or high dose (1.5 x 10(11) vector genomes) for up to 2 years. FINDINGS AAV2-hRPE65v2 was well tolerated and all patients showed sustained improvement in subjective and objective measurements of vision (ie, dark adaptometry, pupillometry, electroretinography, nystagmus, and ambulatory behaviour). Patients had at least a 2 log unit increase in pupillary light responses, and an 8-year-old child had nearly the same level of light sensitivity as that in age-matched normal-sighted individuals. The greatest improvement was noted in children, all of whom gained ambulatory vision. The study is registered with ClinicalTrials.gov, number NCT00516477. INTERPRETATION The safety, extent, and stability of improvement in vision in all patients support the use of AAV-mediated gene therapy for treatment of inherited retinal diseases, with early intervention resulting in the best potential gain. FUNDING Center for Cellular and Molecular Therapeutics at the Childrens Hospital of Philadelphia, Foundation Fighting Blindness, Telethon, Research to Prevent Blindness, F M Kirby Foundation, Mackall Foundation Trust, Regione Campania Convenzione, European Union, Associazione Italiana Amaurosi Congenita di Leber, Fund for Scientific Research, Fund for Research in Ophthalmology, and National Center for Research Resources.


Molecular Therapy | 2010

Gene Therapy for Leber's Congenital Amaurosis is Safe and Effective Through 1.5 Years After Vector Administration

Francesca Simonelli; Albert M. Maguire; Francesco Testa; Eric A. Pierce; Federico Mingozzi; Jeannette L. Bennicelli; Settimio Rossi; Kathleen Marshall; Sandro Banfi; Enrico Maria Surace; Junwei Sun; T. Michael Redmond; Xiaosong Zhu; Kenneth S. Shindler; Gui-shuang Ying; Carmela Ziviello; Carmela Acerra; J. Fraser Wright; Jennifer Wellman McDonnell; Katherine A. High; Jean Bennett; Alberto Auricchio

The safety and efficacy of gene therapy for inherited retinal diseases is being tested in humans affected with Lebers congenital amaurosis (LCA), an autosomal recessive blinding disease. Three independent studies have provided evidence that the subretinal administration of adeno-associated viral (AAV) vectors encoding RPE65 in patients affected with LCA2 due to mutations in the RPE65 gene, is safe and, in some cases, results in efficacy. We evaluated the long-term safety and efficacy (global effects on retinal/visual function) resulting from subretinal administration of AAV2-hRPE65v2. Both the safety and the efficacy noted at early timepoints persist through at least 1.5 years after injection in the three LCA2 patients enrolled in the low dose cohort of our trial. A transient rise in neutralizing antibodies to AAV capsid was observed but there was no humoral response to RPE65 protein. The persistence of functional amelioration suggests that AAV-mediated gene transfer to the human retina does not elicit immunological responses which cause significant loss of transduced cells. The persistence of physiologic effect supports the possibility that gene therapy may influence LCA2 disease progression. The safety of the intervention and the stability of the improvement in visual and retinal function in these subjects support the use of AAV-mediated gene augmentation therapy for treatment of inherited retinal diseases.


Molecular and Cellular Biology | 1995

The PAX3-FKHR fusion protein created by the t(2;13) translocation in alveolar rhabdomyosarcomas is a more potent transcriptional activator than PAX3.

William J. Fredericks; Naomi Galili; Sunil Mukhopadhyay; Giovanni Rovera; Jeannette L. Bennicelli; Frederic G. Barr; Frank J. Rauscher

Alveolar rhabdomyosarcomas are pediatric solid tumors with a hallmark cytogenetic abnormality: translocation of chromosomes 2 and 13 [t(2;13) (q35;q14)]. The genes on each chromosome involved in this translocation have been identified as the transcription factor-encoding genes PAX3 and FKHR. The NH2-terminal paired box and homeodomain DNA-binding domains of PAX3 are fused in frame to COOH-terminal regions of the chromosome 13-derived FKHR gene, a novel member of the forkhead DNA-binding domain family. To determine the role of the fusion protein in transcriptional regulation and oncogenesis, we identified the PAX3-FKHR fusion protein and characterized its function(s) as a transcription factor relative to wild-type PAX3. Antisera specific to PAX3 and FKHR were developed and used to examine PAX3 and PAX3-FKHR expression in tumor cell lines. Sequential immunoprecipitations with anti-PAX3 and anti-FKHR sera demonstrated expression of a 97-kDa PAX3-FKHR fusion protein in the t(2;13)-positive rhabdomyosarcoma Rh30 cell line and verified that a single polypeptide contains epitopes derived from each protein. The PAX3-FKHR protein was localized to the nucleus in Rh30 cells, as was wild-type PAX3, in t(2;13)-negative A673 cells. In gel shift assays using a canonical PAX binding site (e5 sequence), we found that DNA binding of PAX3-FKHR was significantly impaired relative to that of PAX3 despite the two proteins having identical PAX DNA-binding domains. However, the PAX3-FKHR fusion protein was a much more potent transcriptional activator than PAX3 as determined by transient cotransfection assays using e5-CAT reporter plasmids. The PAX3-FKHR protein may function as an oncogenic transcription factor by enhanced activation of normal PAX3 target genes.


Science Translational Medicine | 2012

AAV2 Gene Therapy Readministration in Three Adults with Congenital Blindness

Jean Bennett; Manzar Ashtari; Jennifer Wellman; Kathleen Marshall; Laura Cyckowski; Daniel C. Chung; Sarah McCague; Eric A. Pierce; Yong Chen; Jeannette L. Bennicelli; Xiaosong Zhu; Gui-shuang Ying; Junwei Sun; John Fraser Wright; Alberto Auricchio; Francesca Simonelli; Kenneth S. Shindler; Federico Mingozzi; Katherine A. High; Albert M. Maguire

Repeat administration of gene therapy to the contralateral retina of three congenitally blind patients was safe and resulted in improved vision. Shining a Light with Gene Therapy Gene therapy has great potential for treating certain diseases by providing therapeutic genes to target cells. Administration of a gene therapy vector carrying the RPE65 gene in 12 patients with congenital blindness due to RPE65 mutations led to improvements in retinal and visual function and proved to be a safe and stable procedure. In a follow-up study, the same group of researchers led by Jean Bennett set out to discover whether it would be possible to safely administer the vector and the therapeutic transgene to the contralateral eye of the patients. A big concern was whether the first gene therapy injection might have primed the patients’ immune system to respond to the adeno-associated virus (AAV) vector or the product of the therapeutic transgene that it had delivered. To test the safety and efficacy of a second administration of gene therapy to the second eye, the authors demonstrated that readministration was both safe and effective in animal models. Then, they selected 3 of the original 12 patients and readministered the AAV vector and its RPE65 transgene to the contralateral eye. They assessed safety by evaluating inflammatory responses, immune reactions, and extraocular exposure to the AAV vector. Efficacy was assessed through qualitative and quantitative measures of retinal and visual function including the ability to read letters, the extent of side vision, light sensitivity, the pupillary light reflex, the ability to navigate in dim light, and evidence from neuroimaging studies of cortical activation (which demonstrated that signals from the retina were recognized by the brain). The researchers did not discover any safety concerns and did not identify harmful immune responses to the vector or the transgene product. Before and after comparisons of psychophysical data and cortical responses provided the authors with evidence that gene therapy readministration was effective and mediated improvements in retinal and visual function in the three patients. The researchers report that the lack of immune response and the robust safety profile in this readministration gene therapy study may be due in part to the immune-privileged nature of the eye, and the low dose and very pure preparation of the AAV vector. Demonstration of safe and stable reversal of blindness after a single unilateral subretinal injection of a recombinant adeno-associated virus (AAV) carrying the RPE65 gene (AAV2-hRPE65v2) prompted us to determine whether it was possible to obtain additional benefit through a second administration of the AAV vector to the contralateral eye. Readministration of vector to the second eye was carried out in three adults with Leber congenital amaurosis due to mutations in the RPE65 gene 1.7 to 3.3 years after they had received their initial subretinal injection of AAV2-hRPE65v2. Results (through 6 months) including evaluations of immune response, retinal and visual function testing, and functional magnetic resonance imaging indicate that readministration is both safe and efficacious after previous exposure to AAV2-hRPE65v2.


Cancer Research | 1985

Characteristics of Cultured Human Melanocytes Isolated from Different Stages of Tumor Progression

Meenhard Herlyn; Jan Thurin; Gloria Balaban; Jeannette L. Bennicelli; Dorothee Herlyn; David E. Elder; Edward E. Bondi; DuPont Guerry; Peter C. Nowell; Wallace H. Clark; Hilary Koprowski

Normal melanocytes and melanocytes of normal nevi, primary melanoma in the radial (RGP) and vertical (VGP) growth phases, and metastatic melanoma exhibited and maintained phenotypic differences when grown in tissue culture or in experimental animals. Only metastatic and VGP primary melanoma cells were tumorigenic in athymic nude mice and had nonrandom chromosomal abnormalities involving chromosomes 1, 6, and 7. The colony-forming efficiency in soft agar was also highest in these two cell types. A cell line of RGP primary melanoma had characteristics of both benign and malignant cells: nevus-like morphology; nontumorigenicity in nude mice; but karyotypic abnormality of chromosome 6. It also had a ganglioside pattern similar to that of normal melanocytes but not melanomas, i.e., a high GM3 ganglioside content compared to the amounts of GM2, GD2, and GD3 gangliosides. Binding of monoclonal antibodies secreted by hybridomas generated by immunization of mice with VGP primary and metastatic melanoma was highest with cells and supernatants of cultures from advanced melanoma and least with nevus cells. There was no binding to normal melanocytes except with the monoclonal antibodies specific for nerve growth factor receptor or 9-O-acetyl-GD3 ganglioside. On the other hand, monoclonal anti-nevus antibodies bound to melanocytes, nevus cells, and RGP primary melanoma cells but not to VGP primary or metastatic melanoma cells. Cultured human melanocytic cells appear to be a unique model for the study of tumor progression.


Molecular Therapy | 2008

Reversal of Blindness in Animal Models of Leber Congenital Amaurosis Using Optimized AAV2-mediated Gene Transfer

Jeannette L. Bennicelli; John Fraser Wright; András M. Komáromy; Jonathan B. Jacobs; Bernd Hauck; Olga Zelenaia; Federico Mingozzi; Daniel Hui; Daniel C. Chung; Tonia S. Rex; Zhangyong Wei; Guang Qu; Shangzhen Zhou; Caroline J. Zeiss; Valder R. Arruda; Gregory M. Acland; L. F. Dell'Osso; Katherine A. High; Albert M. Maguire; Jean Bennett

We evaluated the safety and efficacy of an optimized adeno-associated virus (AAV; AAV2.RPE65) in animal models of the RPE65 form of Leber congenital amaurosis (LCA). Protein expression was optimized by addition of a modified Kozak sequence at the translational start site of hRPE65. Modifications in AAV production and delivery included use of a long stuffer sequence to prevent reverse packaging from the AAV inverted-terminal repeats, and co-injection with a surfactant. The latter allows consistent and predictable delivery of a given dose of vector. We observed improved electroretinograms (ERGs) and visual acuity in Rpe65 mutant mice. This has not been reported previously using AAV2 vectors. Subretinal delivery of 8.25 x 10(10) vector genomes in affected dogs was well tolerated both locally and systemically, and treated animals showed improved visual behavior and pupillary responses, and reduced nystagmus within 2 weeks of injection. ERG responses confirmed the reversal of visual deficit. Immunohistochemistry confirmed transduction of retinal pigment epithelium cells and there was minimal toxicity to the retina as judged by histopathologic analysis. The data demonstrate that AAV2.RPE65 delivers the RPE65 transgene efficiently and quickly to the appropriate target cells in vivo in animal models. This vector holds great promise for treatment of LCA due to RPE65 mutations.


The Lancet | 2016

Safety and durability of effect of contralateral-eye administration of AAV2 gene therapy in patients with childhood-onset blindness caused by RPE65 mutations: a follow-on phase 1 trial

Jean Bennett; Jennifer Wellman; Kathleen Marshall; Sarah McCague; Manzar Ashtari; Julie DiStefano-Pappas; Okan Elci; Daniel C. Chung; Junwei Sun; J. Fraser Wright; Dominique Cross; Puya Aravand; Laura Cyckowski; Jeannette L. Bennicelli; Federico Mingozzi; Alberto Auricchio; Eric A. Pierce; Jason Ruggiero; Bart P. Leroy; Francesca Simonelli; Katherine A. High; Albert M. Maguire

BACKGROUND Safety and efficacy have been shown in a phase 1 dose-escalation study involving a unilateral subretinal injection of a recombinant adeno-associated virus (AAV) vector containing the RPE65 gene (AAV2-hRPE65v2) in individuals with inherited retinal dystrophy caused by RPE65 mutations. This finding, along with the bilateral nature of the disease and intended use in treatment, prompted us to determine the safety of administration of AAV2-hRPE65v2 to the contralateral eye in patients enrolled in the phase 1 study. METHODS In this follow-on phase 1 trial, one dose of AAV2-hRPE65v2 (1.5 × 10(11) vector genomes) in a total volume of 300 μL was subretinally injected into the contralateral, previously uninjected, eyes of 11 children and adults (aged 11-46 years at second administration) with inherited retinal dystrophy caused by RPE65 mutations, 1.71-4.58 years after the initial subretinal injection. We assessed safety, immune response, retinal and visual function, functional vision, and activation of the visual cortex from baseline until 3 year follow-up, with observations ongoing. This study is registered with ClinicalTrials.gov, number NCT01208389. FINDINGS No adverse events related to the AAV were reported, and those related to the procedure were mostly mild (dellen formation in three patients and cataracts in two). One patient developed bacterial endophthalmitis and was excluded from analyses. We noted improvements in efficacy outcomes in most patients without significant immunogenicity. Compared with baseline, pooled analysis of ten participants showed improvements in mean mobility and full-field light sensitivity in the injected eye by day 30 that persisted to year 3 (mobility p=0.0003, white light full-field sensitivity p<0.0001), but no significant change was seen in the previously injected eyes over the same time period (mobility p=0.7398, white light full-field sensitivity p=0.6709). Changes in visual acuity from baseline to year 3 were not significant in pooled analysis in the second eyes or the previously injected eyes (p>0.49 for all time-points compared with baseline). INTERPRETATION To our knowledge, AAV2-hRPE65v2 is the first successful gene therapy administered to the contralateral eye. The results highlight the use of several outcome measures and help to delineate the variables that contribute to maximal benefit from gene augmentation therapy in this disease. FUNDING Center for Cellular and Molecular Therapeutics at The Childrens Hospital of Philadelphia, Spark Therapeutics, US National Institutes of Health, Foundation Fighting Blindness, Institute for Translational Medicine and Therapeutics, Research to Prevent Blindness, Center for Advanced Retinal and Ocular Therapeutics, Mackall Foundation Trust, F M Kirby Foundation, and The Research Foundation-Flanders.


Science Translational Medicine | 2010

Safety and Efficacy of Subretinal Readministration of a Viral Vector in Large Animals to Treat Congenital Blindness

Defne Amado; Federico Mingozzi; Daniel Hui; Jeannette L. Bennicelli; Zhangyong Wei; Yifeng Chen; Erin Bote; Rebecca Grant; Jeffrey A. Golden; Kristina Narfström; Nasreen A. Syed; Stephen E. Orlin; Katherine A. High; Albert M. Maguire; Jean Bennett

After successful gene therapy to correct retinal degeneration in one eye, treatment of the second eye is safe and effective—even when immunity to the vector is present—in nonhuman primates and dogs. Both Sides Now Blind and deaf from birth, Helen Keller—whose life is the subject of the drama The Miracle Worker—once remarked that “the world is full of suffering; it is also full of overcoming.” Ms. Keller knew something about rising above one’s circumstances—despite her dual disability, she became a renowned author, activist, and lecturer. Last year, through a ground-breaking clinical trial, a research team from The Children’s Hospital of Philadelphia (CHOP) and the University of Pennsylvania helped a group of children to overcome their near-complete blindness by a different route—a gene therapy regimen that replaced a mutated gene and partially restored visual function in one eye of all 12 patients. Now, this same team of researchers has taken an essential step toward the ability to offer these patients the same sight-restoring treatment for their second eyes. These trial participants had all inherited a specific type of Leber’s congenital amaurosis (LCA), a class of progressive retinal degenerative conditions that can be detected in infancy and results in complete blindness by the age of 40. The loss of photosensitive retinal cells characteristic of this class of diseases stems from mutations in one of 14 genes. The LCA-RPE65 patients in the CHOP trial all had aberrant copies of the RPE65 gene, which encodes an enzyme in the retinal pigment epithelium whose function is a crucial component of the visual cycle. Therefore, surgeons delivered a wild-type copy of RPE65 housed in an adeno-associated virus vector (AAV2-hRPE65v2) into one of the eyes of each of these trial subjects. As one might imagine, the sight-restoring success of this gene therapy trial has patients clamoring for more—more treatment, that is, of their second eyes. But first, researchers needed to assess whether a second treatment would be safe and effective. Specifically, scientists investigated whether subjects who developed neutralizing antibodies (NAbs) directed against the AAV vector used in the first round of gene therapy would benefit from a second round or whether the anti-AAV NAbs would impede gene transfer. This week’s issue of Science Translational Medicine reports the results of Amado et al. on the immunological and functional consequences of serial subretinal readministration of the RPE65-carrying AAV vector. The authors performed their studies in two large animal models—a canine model of LCA-RPE65 and nonhuman primates without eye disease. Both animals sport eyes similar in size to those of humans, and all primates have macula, a specialized region of the retina responsible for central vision. Also, because nonhuman primates can be infected with wild-type AAV and develop antibodies to viral components, these animals likely give researchers a window into the human immune response that manifests upon vector injection. The authors show that the readministration treatment is safe and effective in both models, even in animals that display preexisting immunity to the vector. These results suggest that a patient who has undergone AAV2-hRPE65v2—mediated gene therapy in one eye may indeed enjoy success from a second surgery. Furthermore, scientists had routinely excluded from clinical trials patients who already carried NAbs to AAVs in their sera. And, as Amado et al. also show, this crowd encompasses a substantial portion of the human population, a number that increases with age. Now, LCA-RPE65 patients with antibodies to AAV may be eligible for gene therapy. This study thus represents an essential step in the further translation of gene therapy for LCA—and a reason for celebration in the gene therapy research community. Leber’s congenital amaurosis (LCA) is a group of severe inherited retinal degenerations that are symptomatic in infancy and lead to total blindness in adulthood. Recent clinical trials using recombinant adeno-associated virus serotype 2 (rAAV2) successfully reversed blindness in patients with LCA caused by RPE65 mutations after one subretinal injection. However, it was unclear whether treatment of the second eye in the same manner would be safe and efficacious, given the potential for a complicating immune response after the first injection. Here, we evaluated the immunological and functional consequences of readministration of rAAV2-hRPE65v2 to the contralateral eye using large animal models. Neither RPE65-mutant (affected; RPE65−/−) nor unaffected animals developed antibodies against the transgene product, but all developed neutralizing antibodies against the AAV2 capsid in sera and intraocular fluid after subretinal injection. Cell-mediated immune responses were benign, with only 1 of 10 animals in the study developing a persistent T cell immune response to AAV2, a response that was mediated by CD4+ T cells. Sequential bilateral injection caused minimal inflammation and improved visual function in affected animals. Thus, subretinal readministration of rAAV2 in animals is safe and effective, even in the setting of preexisting immunity to the vector, a parameter that has been used to exclude patients from gene therapy trials.


Laboratory Investigation | 2004

Inducible short-term and stable long-term cell culture systems reveal that the PAX3-FKHR fusion oncoprotein regulates CXCR4, PAX3, and PAX7 expression

Oana Tomescu; Shujuan J. Xia; Donna Strezlecki; Jeannette L. Bennicelli; Jill P. Ginsberg; Bruce R. Pawel; Frederic G. Barr

In the pediatric cancer alveolar rhabdomyosarcoma (ARMS), the 2;13 chromosomal translocation juxtaposes the PAX3 and FKHR genes to generate a chimeric transcription factor. To explore molecular pathways altered by this oncoprotein, we generated an inducible form by fusing PAX3-FKHR to a modified estrogen receptor ligand-binding domain and expressed this construct in the RD embryonal rhabdomyosarcoma cell line. This inducible system permits short-term evaluation of downstream expression targets of PAX3-FKHR and complements a panel of stable long-term RD subclones constitutively expressing PAX3-FKHR. Using these two sets of resources, we investigated several candidate PAX3-FKHR target genes. First, we demonstrated in both short-term and long-term systems that PAX3-FKHR upregulates expression of the gene encoding the chemokine receptor CXCR4. In addition, we found that expression of wild-type PAX3 is upregulated, whereas expression of wild-type PAX7 is downregulated by PAX3-FKHR. In the presence of cycloheximide, CXCR4 and PAX3 are still inducible, supporting the hypothesis that these genes are direct transcriptional targets of PAX3-FKHR. Finally, studies of ARMS tumors revealed CXCR4, PAX3, and PAX7 expression levels consistent with our cell culture results. These findings of genes regulated by PAX3-FKHR will direct future biological and clinical investigation to important pathways contributing to ARMS tumorigenesis and progression.

Collaboration


Dive into the Jeannette L. Bennicelli's collaboration.

Top Co-Authors

Avatar

Jean Bennett

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Albert M. Maguire

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Frederic G. Barr

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Federico Mingozzi

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Junwei Sun

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

DuPont Guerry

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Katherine A. High

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Eric A. Pierce

Massachusetts Eye and Ear Infirmary

View shared research outputs
Researchain Logo
Decentralizing Knowledge